Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 6 de 6
Filter
Add more filters










Database
Language
Publication year range
1.
Br J Cancer ; 123(10): 1481-1489, 2020 11.
Article in English | MEDLINE | ID: mdl-32839491

ABSTRACT

BACKGROUND: AsiDNA, a first-in-class oligonucleotide-mimicking double-stranded DNA breaks, acts as a decoy agonist to DNA damage response in tumour cells. It also activates DNA-dependent protein kinase and poly (adenosine diphosphate [ADP]-ribose) polymerase enzymes that induce phosphorylation of H2AX and protein PARylation. METHODS: The aim of this Phase 1 study was to determine dose-limiting toxicities (DLTs), maximum tolerated dose (MTD), safety and pharmacokinetics/pharmacodynamics of AsiDNA administered daily for 3 days in the first week then weekly thereafter. Twenty-two patients with advanced solid tumours were enrolled in 5 dose levels: 200, 400, 600, 900, and 1300 mg, using a 3 + 3 design. RESULTS: The MTD was not reached. IV AsiDNA was safe. Two DLTs (grade 4 and grade 3 hepatic enzymes increased at 900 and 1300 mg), and two related SAE at 900 mg (grade 3 hypotension and grade 4 hepatic enzymes increased) were reported. AsiDNA PK increased proportionally with dose. A robust activation of DNA-PK by a significant posttreatment increase of γH2AX was evidenced in tumour biopsies. CONCLUSION: The dose of 600 mg was identified as the optimal dose for further clinical development. CLINICAL TRIAL REGISTRATION: Clinical trial registration (NCT number): NCT03579628.


Subject(s)
Cholesterol/analogs & derivatives , DNA/administration & dosage , DNA/adverse effects , DNA/pharmacokinetics , Neoplasms/drug therapy , Administration, Intravenous , Adult , Aged , Belgium , Cholesterol/administration & dosage , Cholesterol/adverse effects , Cholesterol/pharmacokinetics , DNA Repair/drug effects , Disease Progression , Dose-Response Relationship, Drug , Female , France , Humans , Male , Maximum Tolerated Dose , Middle Aged , Neoplasms/metabolism , Neoplasms/pathology
2.
Cancer Immunol Immunother ; 68(3): 379-393, 2019 Mar.
Article in English | MEDLINE | ID: mdl-30547218

ABSTRACT

Immune checkpoint inhibitors are currently tested in different combinations in patients with advanced hepatocellular carcinoma (HCC). Nivolumab, an anti-PD-1 agent, has gained approval in the second-line setting in the USA. Epigenetic drugs have immune-mediated antitumor effects that may improve the activity of immunotherapy agents. Our aim was to study the therapeutic efficacy of checkpoint inhibitors (anti-CTLA-4 and anti-PD-1 antibodies) in combination with the histone deacetylase inhibitor (HDACi) Belinostat. In a subcutaneous Hepa129 murine HCC model, we demonstrated that Belinostat improves the antitumor activity of anti-CTLA-4 but not of anti-PD-1 therapy. This effect correlated with enhanced IFN-γ production by antitumor T-cells and a decrease in regulatory T-cells. Moreover, the combination induced early upregulation of PD-L1 on tumor antigen-presenting cells and late expression of PD-1 on tumor-infiltrating effector T-cells, suggesting the suitability of PD-1 blockade. Indeed, Belinostat combined with the simultaneous blockade of CTLA-4 and PD-1 led to complete tumor rejection. These results provide a rationale for testing Belinostat in combination with checkpoint inhibitors to enhance their therapeutic activity in patients with HCC.


Subject(s)
CTLA-4 Antigen/antagonists & inhibitors , Carcinoma, Hepatocellular/drug therapy , Histone Deacetylase Inhibitors/administration & dosage , Hydroxamic Acids/administration & dosage , Liver Neoplasms, Experimental/drug therapy , Programmed Cell Death 1 Receptor/antagonists & inhibitors , Sulfonamides/administration & dosage , Animals , Cell Line, Tumor , Disease Models, Animal , Drug Therapy, Combination , Female , Humans , Macrophages/physiology , Mice , Mice, Inbred C3H , T-Lymphocytes, Regulatory/immunology
3.
Lancet ; 377(9783): 2093-102, 2011 Jun 18.
Article in English | MEDLINE | ID: mdl-21652063

ABSTRACT

BACKGROUND: Young people aged 10-24 years represent 27% of the world's population. Although important health problems and risk factors for disease in later life emerge in these years, the contribution to the global burden of disease is unknown. We describe the global burden of disease arising in young people and the contribution of risk factors to that burden. METHODS: We used data from WHO's 2004 Global Burden of Disease study. Cause-specific disability-adjusted life-years (DALYs) for young people aged 10-24 years were estimated by WHO region on the basis of available data for incidence, prevalence, severity, and mortality. WHO member states were classified into low-income, middle-income, and high-income countries, and into WHO regions. We estimated DALYs attributable to specific global health risk factors using the comparative risk assessment method. DALYs were divided into years of life lost because of premature mortality (YLLs) and years lost because of disability (YLDs), and are presented for regions by sex and by 5-year age groups. FINDINGS: The total number of incident DALYs in those aged 10-24 years was about 236 million, representing 15·5% of total DALYs for all age groups. Africa had the highest rate of DALYs for this age group, which was 2·5 times greater than in high-income countries (208 vs 82 DALYs per 1000 population). Across regions, DALY rates were 12% higher in girls than in boys between 15 and 19 years (137 vs 153). Worldwide, the three main causes of YLDs for 10-24-year-olds were neuropsychiatric disorders (45%), unintentional injuries (12%), and infectious and parasitic diseases (10%). The main risk factors for incident DALYs in 10-24-year-olds were alcohol (7% of DALYs), unsafe sex (4%), iron deficiency (3%), lack of contraception (2%), and illicit drug use (2%). INTERPRETATION: The health of young people has been largely neglected in global public health because this age group is perceived as healthy. However, opportunities for prevention of disease and injury in this age group are not fully exploited. The findings from this study suggest that adolescent health would benefit from increased public health attention. FUNDING: None.


Subject(s)
Developing Countries/statistics & numerical data , Health Status , Adolescent , Africa/epidemiology , Asia, Southeastern/epidemiology , Child , Cost of Illness , Developed Countries/statistics & numerical data , Female , Humans , Male , Morbidity , Mortality , Quality-Adjusted Life Years , Risk Factors , World Health Organization , Young Adult
4.
J Clin Invest ; 121(1): 410-21, 2011 Jan.
Article in English | MEDLINE | ID: mdl-21135506

ABSTRACT

Dysregulated angiogenesis is a hallmark of chronic inflammatory diseases, including psoriasis, a common skin disorder that affects approximately 2% of the population. Studying both human psoriasis in 2 complementary xenotransplantation models and psoriasis-like skin lesions in transgenic mice with epidermal expression of human TGF-ß1, we have demonstrated that antiangiogenic non-viral somatic gene therapy reduces the cutaneous microvasculature and alleviates chronic inflammatory skin disorders. Transient muscular expression of the recombinant disintegrin domain (RDD) of metargidin (also known as ADAM-15) by in vivo electroporation reduced cutaneous angiogenesis and vascularization in all 3 models. As demonstrated using red fluorescent protein-coupled RDD, the treatment resulted in muscular expression of the gene product and its deposition within the cutaneous hyperangiogenic connective tissue. High-resolution ultrasound revealed reduced cutaneous blood flow in vivo after electroporation with RDD but not with control plasmids. In addition, angiogenesis- and inflammation-related molecular markers, keratinocyte proliferation, epidermal thickness, and clinical disease scores were downregulated in all models. Thus, non-viral antiangiogenic gene therapy can alleviate psoriasis and may do so in other angiogenesis-related inflammatory skin disorders.


Subject(s)
Genetic Therapy , Neovascularization, Pathologic/therapy , Psoriasis/therapy , ADAM Proteins/genetics , Animals , Disease Models, Animal , Endothelial Cells/physiology , Female , Gene Expression , Humans , In Vitro Techniques , Male , Membrane Proteins/genetics , Mice , Mice, Transgenic , Neovascularization, Pathologic/genetics , Neovascularization, Pathologic/pathology , Neovascularization, Pathologic/physiopathology , Psoriasis/genetics , Psoriasis/pathology , Psoriasis/physiopathology , Recombinant Fusion Proteins/genetics , Transplantation, Heterologous
5.
Cancer Res ; 64(6): 2062-9, 2004 Mar 15.
Article in English | MEDLINE | ID: mdl-15026344

ABSTRACT

Metargidin, a transmembrane protein of the adamalysin family, and integrins, e.g., alpha5beta1 and alphav, are preferentially expressed on endothelial cells on angiogenesis. Furthermore, metargidin interacts with these integrins via its disintegrin domain. In this study, recombinant human disintegrin domain (RDD) was produced in Escherichia coli by subcloning its cDNA into the pGEX-2T vector, and the effect of purified RDD on different steps of angiogenesis was evaluated. At concentrations of 2-10 micro g/ml, RDD exhibited inhibitory activities in a variety of in vitro functional assays, including endothelial cell proliferation and adhesion on the integrin substrates fibronectin, vitronectin, and fibrinogen. RDD (10 micro g/ml) totally abrogated endothelial cell migration and blocked most capillary formation in a three-dimensional fibrin gel. To test RDD efficacy in vivo, the RDD gene inserted into pBi vector containing a tetracycline-inducible promoter was electrotransferred into nude mouse muscle. RDD was successfully synthesized by muscle cells in vivo as shown by immunolabeling and Western blotting. In addition, 78% less MDA-MB-231 tumor growth, associated with strong inhibition of tumor angiogenesis, was observed in athymic mice bearing electrotransferred RDD. Moreover, in the presence of RDD, 74% fewer B16F10 melanoma lung metastases were found in C57BL/6 mice. Taken together, these results identified this RDD as a potent intrinsic inhibitor of angiogenesis, tumor growth, and metastasis, making it a promising tool for use in anticancer treatment.


Subject(s)
Antineoplastic Agents/therapeutic use , Disintegrins/therapeutic use , Lung Neoplasms/prevention & control , Melanoma, Experimental/prevention & control , Membrane Proteins/therapeutic use , Metalloendopeptidases/therapeutic use , Neovascularization, Pathologic/prevention & control , ADAM Proteins , Animals , Apoptosis/drug effects , Cell Adhesion/drug effects , Cell Division/drug effects , Cell Movement/drug effects , Dose-Response Relationship, Drug , Endothelium, Vascular/drug effects , Endothelium, Vascular/pathology , Escherichia coli/genetics , Female , Lung Neoplasms/blood supply , Lung Neoplasms/secondary , Melanoma, Experimental/blood supply , Melanoma, Experimental/secondary , Mice , Mice, Inbred C57BL , Mice, Nude , Muscle, Skeletal/pathology , Recombinant Proteins/therapeutic use , Tumor Cells, Cultured
6.
Mol Ther ; 8(3): 425-33, 2003 Sep.
Article in English | MEDLINE | ID: mdl-12946315

ABSTRACT

We describe an approach employing intramuscular plasmid electrotransfer to deliver secretable forms of K1-5 and K1-3-HSA (a fusion of K1-3 with human serum albumin), which span, respectively, five and three of the five kringle domains of plasminogen. A tetracycline-inducible system (Tet-On) composed of three plasmids coding, respectively, for the transgene, the tetracycline transcriptional activator rtTA, and the silencer tTS was employed. K1-3-HSA and K1-5, produced from C2C12 muscle cells, were found to inhibit endothelial cell (HMEC-1) proliferation by 30 and 51%, respectively. In vivo, the expression of the transgene upon doxycycline stimulation was rapid, stable, and tightly regulated (no background expression) and could be maintained for at least 3 months. Blood half-lives of 2.1 and 3.7 days were found for K1-5 and K1-3-HSA, respectively. The K1-5 protein was secreted from muscle into blood at a level of 45 ng/ml, which was sufficient to inhibit MDA-MB-231 tumor growth by 81% in nude mice and B16-F10 melanoma cell lung invasion in C57BL/6 mice by 73%. PECAM-1 immunostaining studies revealed modest tumor vasculature in mice expressing K1-5. In contrast, K1-3-HSA, although secreted into blood at much higher level (250 ng/ml) than K1-5, had no effect on tumor growth.


Subject(s)
Angiogenesis Inhibitors/genetics , Electroporation , Genetic Vectors , Muscle, Skeletal/metabolism , Neoplasms/therapy , Angiogenesis Inhibitors/metabolism , Gene Expression Regulation , Neoplasm Metastasis/therapy , Peptides/genetics , Peptides/metabolism , Plasmids , Plasminogen/genetics , Plasminogen/metabolism , Tetracycline/metabolism , Time Factors
SELECTION OF CITATIONS
SEARCH DETAIL
...